What is the correct use of established clotting factors, prothrombin complex concentrates (PCCs), and activated factor VII in bleeding complications of trauma, surgery, and old and new oral anticoagulants? How will new clotting factors, specifically the long-acting factors, change the hemostatic management of coagulation deficiency disorders? From bench to bedside, comparative coagulation studies and clinical trials of modified clotting factors are providing insights to help guide hemostatic management of congenital and acquired bleeding disorders. Comparative thrombin-generation studies and preclinical and clinical trials suggest that PCCs and fresh-frozen plasma are effective in reversing the anticoagulant effects of warfarin, yet there are few data to guide reversal of the new oral anticoagulants dabigatran and rivaroxaban. Although coagulation studies support the use of PCCs to reverse new oral anticoagulants, correlation with clinical response is variable and clinical trials in bleeding patients are needed. For congenital bleeding disorders, exciting new technologies are emerging from the bench. Data from clinical trials of molecularly modified coagulation factors with extended half-lives suggest the possibility of fewer infusions, reduced bleeds, and better quality of life in persons with hemophilia. Preclinical studies of other novel prohemostatic approaches for hemophilia and other congenital coagulation disorders include RNA interference silencing of antithrombin, monoclonal anti-tissue factor pathway inhibitor (anti-antibody, anti-tissue factor pathway inhibitor) aptamer, bispecific anti-IXa/X antibody, and fucoidans. Understanding the comparative coagulation studies of established prohemostatic agents, the pharmacokinetics of new long-acting clotting factors, and their correlation with bleeding outcomes will provide opportunities to optimize the hemostatic management of both congenital and acquired hemostatic disorders.

Background

Hemostatic management of patients with excessive bleeding in trauma, surgery, and with oral anticoagulant therapy continues to be a major challenge. Increasing evidence from the bench and bedside indicates that prohemostatic agents such as prothrombin complex concentrates (PCCs), factor eight inhibitor bypass activity (FEIBA), and activated factor VII (rFVIIa) improve quality and quantity of clot formation and reverse excessive bleeding. Current recommendations for surgical/trauma hemostasis and warfarin reversal are based on data from comparative coagulation studies and preclinical and clinical trials, yet few data exist regarding new oral thrombin and Xa inhibitors and clinical trials on these are not available. The purpose of this update is to review current laboratory and clinical studies of prohemostatic agents to guide hemostatic management of excess bleeding in trauma, surgery, and anticoagulant reversal.

Whether induced by trauma, surgery, or excess anticoagulation, bleeding occurs through vessel injury. Vessel injury initiates coagulation through its interaction with coagulation factors, the vessel wall, and platelets that ultimately ensures the formation of a lifesaving fibrin clot (Figure 1). The damaged tissue from injured vessels (tissue factor, TF) activates factor VII (fVIIa), triggers a complex series of reactions that successively activate the tenase complex (the conversion of X by IXa with cofactor VIIIa) and the prothrombinase complex (the conversion of II by Xa with factor Va), generating thrombin (IIa; Figure 1). Thrombin activates fibrin clot formation, promotes clot stabilization and cross-linking through activation of fXIII, and provides clot protection through activation of thrombin-activatable fibrinolysis inhibitor. This process can be monitored by qualitative and quantitative laboratory measures of clot formation, specifically the thrombin generation assay (TGA) and thromboelastometry (ROTEM), which provide more comprehensive clot assessment than the prothrombin time (PT) and activated partial thromboplastin time (aPTT), which measure one-dimensional time to clot formation.1  The TGA assay measures the capacity of plasma to form thrombin, including endogenous thrombin potential (ETP; area under the curve) and lag time (LT; the time to initiation of thrombin generation); and ROTEM quantifies the effect of plasma and its cellular components on clot formation, including clotting time, maximum clot firmness, and clot lysis.2  The TGA and ROTEM assays can be used to assess the relative prohemostatic effects of PCCs, FEIBA, and rFVIIa in the management of hemorrhage with surgery, trauma, and excess anticoagulation. The purpose of this section is to review TGA and ROTEM findings and clinical bleeding outcomes after PCCs and rFVIIa in excessive bleeding with surgery, procedures, or anticoagulant reversal (Table 1) and current guidance for their use.

Figure 1.

Coagulation pathway, PCCs and rFVIIa, and TGA and ROTEM assays.

Figure 1.

Coagulation pathway, PCCs and rFVIIa, and TGA and ROTEM assays.

Close modal
Table 1.

Studies of prohemostatic agents for off-label use in surgery, trauma, and anticoagulant reversal

Studies of prohemostatic agents for off-label use in surgery, trauma, and anticoagulant reversal
Studies of prohemostatic agents for off-label use in surgery, trauma, and anticoagulant reversal

CPB indicates cardiopulmonary bypass; ECT, ecarin clotting time; ICH, intracranial hemorrhage; INR, international normalized ratio; NA, not available; NQ, not quantified; TT, thrombin time; FFP, fresh-frozen plasma; and cryo, cryoprecipitate.

PCCs

PCCs are factor concentrates that contain factors II, VII, IX, and X, as well as protein C and S, originally developed to control bleeding in hemophilia patients with inhibitors to factor VIII or IX, to “bypass” factor VIII and factor IX in clot formation; and in hemophilia B patients. The so-called 4-factor PCCs such as Beriplex and Octaplex, which are manufactured outside of the United States, and Kcentra, which was recently licensed in the United States, contain factors II, VII, IX, and X, whereas the 3-factor PCCs such as Bebulin and Profiline contain factors II, IX, and X, but low levels of VII. Activated PCCs such as FEIBA were subsequently developed to increase the content of activated clotting factors in PCCs to enhance hemostasis, specifically the content of VIIa, IIa, and Xa. Although the use of PCCs for other reasons than the control of bleeding in hemophilia, for example, for blood loss or anticoagulation reversal, is considered off-label in the United States, it is a licensed indication in Europe. PCCs have a rapid onset of action (within minutes) and viral inactivation is by filtration, nanofiltration, and either pasteurization or solvent detergent treatment. Potential complications of PCC use include thrombosis and thromboembolic events.

rFVIIa

rFVIIa is a recombinant protein originally licensed to treat or prevent bleeding in patients with hemophilia A or B with inhibitors (antibodies) to factor VIII or IX, in patients with congenital factor VII deficiency, and, in Europe, for use in Glanzann thrombasthenia. The mechanism by which rFVIIa corrects hemophilia A or B inhibitors is as a “bypass” therapy, meaning bypassing VIII or IX, which are unavailable for clot formation (due to the inhibitor), and instead activating coagulation through VIIa. rFVIIa is also widely used off-label for life-threatening bleeding after major surgery or trauma or with oral anticoagulants. The mechanism of rFVIIa action is thought to be by boosting clot formation via the production of a “thrombin burst” through activation of IX, X, and II on the surface of activated platelets. In clinical settings, however, where no congenital factor deficiency exists and thrombin formation is adequate, rFVIIa may actually promote excess thrombin, that is, above-normal thrombin generation, resulting in thromboembolic complications3,4  that may be fatal in up to 27% of patients.5  Therefore, rFVIIa in off-label settings, which constitutes 97% of its use,5  may be dangerous, remains unsupported, and is specifically advised against in Europe. The drug is expensive, its risk-benefit profile is not well understood,4  and there is no validated test to monitor its use.

Comparative coagulation and preclinical studies

In animal coagulopathy models, clot stability is improved and blood loss is reduced after PCCs and, to a lesser extent, rFVIIa.6  In a porcine model of dilutional coagulopathy, PCCs provided better hemostatic control than rFVIIa. After liver laceration, pigs receiving 4-factor PCCs (35 U/kg) or rFVIIa (6 μg/kg) each demonstrated improved ROTEM clot strength, but there was more durable thrombin generation and less blood loss with PCC-treated animals.7  Similarly, in a porcine trauma model induced by standardized spleen injury, pigs receiving 4-factor PCC (35 U/kg) showed stronger, more rapid thrombin generation and clinical hemostasis than pigs receiving rFVIIa (180 μg/kg).8  In ROTEM studies, PCCs were also superior to rFVIIa in protecting against fibrinolysis in V-, VIII-, IX-, or X-deficient plasmas.2,9  ROTEM studies in plasmas with low heparin concentration, typical of cardiac bypass surgery, further revealed that adequate clot stability after rFVIIa required the addition of fibrinogen and aprotinin.2  These preclinical data provide in vitro and in vivo evidence that PCCs are more effective than rFVIIa in restoring thrombin generation and reducing blood loss in surgery or anticoagulation reversal.10 

Clinical studies

Clinical studies of coagulopathic patients undergoing surgery or procedures receiving PCCs or rFVIIa indicate that these patients have reduced blood loss and improved hemostasis (Table 1). In a prospective randomized, controlled trial of 43 patients with international normalized ratio (INR) > 2 requiring anticoagulant reversal for procedures or acute hemorrhage, 4-factor PCCs given at 25, 35, or 50 U/kg for INR < 4, INR 4-6, or INR > 6, respectively, corrected the INR to < 1.3 within 30 minutes in 93% of patients and effectively controlled bleeding.11  In patients undergoing cardiopulmonary bypass, 3-factor PCCs (18.9-30.9 U/kg) reduced transfusion requirement—fresh-frozen plasma (FFP) by 22% and platelet transfusions by 12%—to a greater degree than rFVIIa (90-120 μg/kg).12,13  Among surgical patients with severe perioperative bleeding, 4-factor PCCs (2000-3000 U) corrected INR from 1.7 to 1.4, improved hemoglobin and arterial blood pressure, and reduced surgical bleeding.14 

Several studies indicate that rFVIIa provides rapid INR correction and shortens ROTEM clot formation and TGA lag time, although clinical response may be variable and thrombosis more common. In coagulopathic cardiac bypass patients undergoing aortic repair, treatment with rFVIIa alone (90-120 μg/kg) reduced chest tube drainage and transfusion requirement.13  In contrast, in a randomized controlled trial of 24 warfarin-treated subjects undergoing punch skin biopsy, pretreatment with rFVIIa (80 μg/kg) improved clot formation in TGA and ROTEM assays but failed to reduce blood loss or bleeding duration.3  In a postoperative cardiac surgery study, whereas rFVIIa shortened ROTEM clot formation and thrombin generation, fibrinogen was required to achieve clot stability.2  In a cardiac surgery study in which rFVIIa (40 or 80 μg/kg) reduced blood loss and transfusion requirement, there was an increase in thromboembolic events compared with placebo.15  In fact, a systematic review of 16 randomized clinical trials and 48 observational studies of off-label rFVIIa demonstrated the highest thrombotic complication rate occurred in cardiac surgery and intracranial hemorrhage (ICH) patients.4 

In coagulopathic trauma patients, half of whom were receiving warfarin, 3-factor PCCs (25 U/kg) rapidly corrected the INR and reduced the RBC requirement, but, as with most studies of rFVIIa in trauma, showed no survival benefit.16  A recent retrospective review of 124 severely injured trauma patients requiring massive transfusion, however, found that early use of rFVIIa (120 μg/kg) within 2 hours of injury not only reduced hemorrhage and coagulopathy but also decreased 24-hour and 30-day mortality.17  Despite this finding, controversy remains regarding rFVIIa use given its high cost and lack of dosing guidelines.

A comparison study of 3-factor PCCs (25 U/kg) with rFVIIa (90 μg/kg) in 85 traumatic brain injury patients revealed significantly greater reduction in RBC and FFP requirement and lower mortality rate in the PCC group.18  The combination of rFVIIa (1.0 mg) and 3-factor PCCs to treat 46 patients with intracerebral hemorrhage led to rapid INR reversal that was maintained during hospitalization, with hemorrhagic expansion in only 4 patients.19  In a randomized trial in patients with acute major hemorrhage with surgical procedures, all of whom also received vitamin K, the 4-factor PCC Kcentra at 25 U/kg for INR 2.0-3.9, 35 U/kg for INR 4-6, and 50 U/kg for INR > 6, was significantly superior to FFP, shortening the INR to 1.3 in the first 24 hours, with correction beginning by 30 minutes.20 

Finally, it is noteworthy that the use of ROTEM testing to guide PCC therapy in coagulopathic surgical patients has reduced transfusion requirement, blood loss, ICU admission, and deaths compared with routine monitoring without ROTEM.21 

Comparative coagulation and preclinical studies

The prohemostatic effects of PCCs and FFP have been assessed by in vitro thrombin generation in warfarin-treated whole blood. PCCs were shown to be more effective than FFP in restoring PT/INR and thrombin generation (peak and lag time) and, among the PCCs, 4-factor PCCs were more effective than 3-factor PCCs.22  The degree of PT/INR correction is related to the starting INR: whereas 3- and 4-factor PCCs restore PT/INR in warfarin-treated plasma samples with INR < 3, only 4-factor PCCs correct PT/INR and thrombin generation in factor VII-deficient plasma when the INR is > 10.22  The latter is attributed to the low factor VII content of 3-factor PCCs and is consistent with the improved thrombin generation when FFP is combined with 3-factor PCCs in warfarin-treated plasma with INR > 4.23 

Clinical studies

During warfarin reversal in 60 coagulopathic patients, some requiring emergency surgery, 4-factor PCCs (15-83 U/kg) corrected the INR to 1.4 within 1 hour in 92% of patients (Table 1).24  In 3 randomized trials of warfarin reversal for ICH, 3-factor PCCs (25-50 U/kg) provided significantly faster INR correction than FFP.25  The addition of FFP to 3-factor PCCs improved warfarin reversal response rate: in 40 patients with INR > 5; 50% achieved INR correction with either FFP or 3-factor PCCs (25-50 U/kg) alone, whereas 89% achieved correction with the combination of FFP and 3-factor PCCs.23 

In a study of preoperative warfarin reversal a single dose of 4-factor PCCs (1000-2000 U) provided INR correction from 2.8 to 1.5 with no postoperative bleeding.14  INR correction was more rapid with 3-factor PCCs (4.8 vs 7.3 hours) and also when 4-factor PCCs were combined with FFP (3 vs 8.9 hours) than with FFP alone.12  INR correction with 3-factor PCCs, however, was limited by the initial INR. Although 4-factor PCCs corrected INR despite the starting INR, 3-factor PCCs corrected INR in only 33% of those with INR > 4 and in none of those with a starting INR ≥ 6.11,26,27  In 11 retrospective studies of acute warfarin reversal for acute bleeding, including ICH, rFVIIa (10-90 μg/kg) rapidly corrected the INR but, unlike with PCCs, the reduction in bleeding was equivocal.28  Similar to the greater degree of clinical bleeding response with PCCs compared with rFVIIa, ROTEM clot stability and clot lysis time assays in warfarin-treated patients improved to a greater degree with PCCs than with rFVIIa.29 

Introduction

With efficacy and safety data from clinical trials of new oral anticoagulants (NOACs), the thrombin inhibitor dabigatran and the Xa inhibitors rivaroxaban and apixaban have been increasingly used in the clinical setting. Despite short half-life, short duration of action, no requirement for monitoring, few drug interactions, and lower bleeding rate, reversal of these agents for life-threatening bleeding is challenging because there are no specific antidotes. However, despite laboratory improvement with hemostatic agents in thrombin generation (TGA), LT, and time to peak thrombin, other assays may not correct30  and clinical bleeding may continue unchecked. Therefore, management of bleeding with NOACs is challenging and, in the absence of clinical trials, clinical guidance is based on data from animal models, healthy volunteers, and case reports.

Comparative coagulation and preclinical studies

In a dabigatran-induced murine coagulopathy model after tail transection injury,26  FEIBA improved bleeding time significantly better than PCCs, rFVIIa, or the combination of PCCs and rFVIIa, but none of these interventions reduced blood loss.31  In the dabigatran-induced rabbit coagulopathy model after standardized kidney incision, PCCs in increasing doses of 20, 35, and 50 U/kg incrementally increased peak thrombin generation and speed of thrombus formation and reduced PT, but blood loss was completely normalized only at the highest dose.32  In a third model, the dabigatran-induced murine ICH model induced by striatal collagenase injection, ICH could be prevented by 4-factor PCCs pretreatment (25-100 U/kg), but not rFVIIa (8 mg/kg) or FFP, and reduction in clinical bleeding as measured by 24-hour ICH expansion was only observed with PCCs and, to a lesser extent, with FFP, but not with rFVIIa.33  In rat and baboon continuous bleeding models induced by continuous infusion rivaroxaban, the administration of PCCs (25 or 50 U/kg), aPCC (50 or 100 U/kg), or rVIIa (400 μg/kg) each reversed the anticoagulant effects of rivaroxaban, as measured by PT and thrombin time (TT), but rFVIIa did not correct thrombin antithrombin complexes by TGA and, moreover, none completely corrected coagulation values to baseline.34  Further, there was poor correlation with bleeding response, measured by mesenteric bleeding time, which failed to completely correct, and this was attributed to the slow generation of Xa to neutralize rivaroxaban and completely stop bleeding.34 

Clinical trials

Although there are no clinical trials of thrombin or Xa inhibitor reversal, several randomized trials in healthy volunteers have investigated laboratory reversal of these agents. In volunteers anticoagulated with rivaroxaban, PCCs (50 U/kg) completely reversed rivaroxaban-induced PT prolongation and TT prolongation and improved ETP.30  In contrast, PCCs at the same dose failed to reverse the anticoagulant effect of dabigatran in these volunteers, as measured by aPTT, TT, and ETP.35  In a second study of healthy volunteers anticoagulated with dabigatran or rivaroxaban, thrombin generation correction by FEIBA (20-120 U/kg) was greater than that by 4-factor PCCs (12.5-50 U/kg) or rFVIIa (20-120 μg/kg).30  Although LT and ETP corrected after FEIBA, only LT corrected after PCCs and only LT corrected after rFVIIa.30  Few data exist in bleeding patients about how improvement in clot formation in TGA relates to clinical bleeding response. In a single available case report of dabigatran-associated bleeding in a patient undergoing aortic valve and coronary artery bypass surgery, high-dose rFVIIa (21.6 mg in 5 doses of 2.4 mg or 7.2 mg each) followed by hemodialysis successfully reduced chest and pericardial blood loss.36 

Bleeding in surgery and trauma

PCCs are more effective than rFVIIa or FFP in restoring thrombin generation and/or INR reversal and reducing blood loss in surgery and trauma regardless of whether there has been previous warfarin use.10,11,16,20  In randomized trials, 3-factor PCCs (25-50 U/kg) have demonstrated more rapid INR correction than FFP25  and when FFP is administered with 3-factor PCCs, response rates improve.23  Although the combination of 3-factor PCCs with low-dose rFVIIa is effective in reversing INR19  and rFVIIa may provide earlier correction of coagulopathy,2,3,17  safety issues remain. In addition, given the thrombotic risk associated with each agent alone, recommendations regarding the combination must await randomized trials comparing safety and efficacy in bleeding patients. The 4-factor PCCs provide rapid, sustained correction of INR in coagulopathic surgical and trauma patients with better efficacy than FFP and without the associated fluid overload,11,16,20  but should be avoided in patients with recent thromboembolism (ie, within the previous 3 months).20  Further, the use of PCCs and rFVIIa should be judicious, with INR-based dosing for PCCs and careful monitoring for thrombosis.20 

Warfarin reversal

Few clinical trials have addressed the management of anticoagulation-related bleeding complications. The current 2012 Chest guidelines, which are based on small observational studies, case reports, and expert opinion, suggest that patients with major vitamin K antagonist–associated bleeding should be rapidly reversed with PCCs, FFP, or rFVIIa, with 4-factor PCCs preferred over plasma, supplemented by IV vitamin K (5-10 mg).10  The latter provides rapid onset of action, within 6-8 hours, and thus helps to sustain the response of other products with shorter half-lives.10  Dosing recommendations for 3-factor PCCs, as for the recently Food and Drug Administration (FDA)–approved 4-factor PCCs, for warfarin reversal include 25 U/kg for INR 2.0-3.9, 35 U/kg for INR 4.0-6.0, and 50 U/kg for INR > 6.0.11,37  Whereas rFVIIa is considered an alternative to PCCs, both agents are associated with thrombosis risk, especially in those with previous thrombosis,20  and the lowest effective dose of rFVIIa is not established.

Reversal of NOACs

Whether PCCs or rFVIIa can reverse the anticoagulant effects of thrombin or direct Xa inhibitors is not established. In the absence of clinical trials, recommendations for NOAC reversal in bleeding patients are based on preclinical studies, case reports, and expert opinion. The short half-life and lack of a specific antidote suggest that for urgent reversal, NOACs and antiplatelets agents should be held and transfusion support and surgical hemostasis provided if indicated.38  Dialysis with activated charcoal, if initiated within 2 to 4 hours of dabigatran ingestion, will remove it from the circulation,38-41  but not rivaroxaban because it is highly protein bound. Although FFP, rFVIIa, and PCCs may be considered, studies of these agents in humans to reverse NOACs are lacking, there is little correlation between coagulation assay correction and reversal of bleeding, and dosing and monitoring are not established.30,32,34-36,38-44 

Experimental agents

Several novel antidotes to reverse NOACs are in development and some are in early clinical trials development for reversal of anticoagulants. These include an Xa congener that neutralizes Xa coagulation inhibitor function, but has no in vivo effects.45  Also being studied is a thrombin double mutant, W215A/E217A, which shortens thrombin inhibitor-associated aPTT prolongation thrombin generation in vitro.46  Another novel drug in development is the dabigatran-specific antidote aDabi-Fab. This antidote mimics thrombin structure but not thrombin function in vitro, with 350-fold greater affinity for dabigatran than for thrombin. Studies have demonstrated that aDabi-Fab induces a rapid and prolonged reversal of dabigatran-prolonged TT and aPTT.47  Development of such antidotes, if shown to be safe and effective in early phase studies, are anticipated to be evaluated in later-phase clinical trials. These agents may provide additional options for emergency reversal of NOACs.

Background of long-acting FIX proteins

Long-lasting products are generating a great deal of excitement within the field. The new long-lasting protein technologies represent a potential major advance in hemophilia management. There are 13 ongoing long-acting factor VIII and IX clinical trials listed at www.clinicaltrials.gov involving 12 to 93 sites on 6 continents and 12 to 172 patients. Differences in factor VIII and IX half-life translate into shorter duration products for factor VIII derivatives (for more information, please see the chapter by Shapiro in this publication regarding long-acting factor VIII proteins). Early-phase long-acting FIX studies52-54  and one phase 3 study55  have been completed, and the latter is under FDA review. Given the progress in hemophilia treatment, with replacement clotting factors enabling a lifespan comparable to that in unaffected men, why the interest in long-acting factor proteins? Despite revolutionary advances in safety and purity of factors, the standard of care for prevention of hemophilic joint disease is prophylaxis or 2 or 3 times weekly IV factor infusion, which is invasive, burdensome, and, in young children, requires venous access devices that may be complicated by thrombosis and infection. Therefore, there is a theoretical advantage for clotting factors with longer half-lives because they could result in potentially fewer infusions, less frequent bleeding, fewer venous access devices, and better quality of life.

Technologies of long-acting factor IX proteins

Recombinant factor IX products with longer plasma half-lives than currently commercially available products have been in development over the last decade. The so-called “long-acting” factor IX proteins are composed of standard recombinant proteins either conjugated to hydrophilic polymers such as polyethylene glycol (PEG)52  or linked or fused to albumin56  or the Fc fragment of IgG1.57  PEG prolongs half-life by causing steric hindrance of the factor IX protein, reducing its elimination efficiency and protecting clotting proteins from proteolytic degradation.52  Alternatively, fusion technology prolongs half-life through fusion of the recombinant factor IX to albumin56  or to the Fc fragment of IgG1.57  In the case of Fc fusion protein, the Fc fragment binds to the neonatal Fc receptor, is taken up by endothelial cells or macrophages by pinocytosis and recycled within lysosomes to the plasma membrane, protecting it from lysosomal degradation, and improving its circulation half-life.58  When these long-acting factor IX proteins are physiologically activated, the PEG, albumin, and linker are cleaved,56  leaving the recombinant factor IX, with similar function to standard factor IX in chromogenic and activity assays and, for everything but the albumin protein, similar in immunogenicity assays.

Preclinical and clinical studies

Animal studies demonstrate prolonged half-life in the circulation of these long-acting factor IX proteins,52,56,57  compared with standard recombinant factor IX. Improved survival has been demonstrated for the glycopegylated and Fc fusion factor IX proteins after tail vein transection when the procedure is preceded by long-acting factor IX and longer-lived hemostasis compared with recombinant factor IX.59  Early-phase safety and pharmacokinetic studies in patients with severe factor IX deficiency have demonstrated safety and prolonged activity with half-lives ranging from 82 to 93 hours after single-dose pharmacokinetic assessment of the glycopegylated factor IX, N9-GP,52  the factor IX Fc fusion protein rFIXFc,53  and the albumin fusion factor IX, rIX-FP54  compared with recombinant factor IX (Table 2). In one phase 3 study in patients with severe factor IX deficiency, the long-acting factor IX appears to have comparable efficacy to recombinant factor IX in the treatment and prevention of bleeds, with a prolonged half-life and a factor IX level above 1%, the level below which spontaneous bleeds may occur, up to at least 14 days in 50% of patients.55  Preliminary data show no evidence of inhibitor antibodies, anaphylaxis, or allergy and no drug-related serious adverse events.

Table 2.

Long-acting rFIX clotting proteins

Long-acting rFIX clotting proteins
Long-acting rFIX clotting proteins

For the first time, long-lasting clotting factor therapy provides the possibility of a simpler, more durable treatment regimen for persons with hemophilia. If safety, efficacy, enhanced recovery, and half-life are confirmed in large clinical studies, it is anticipated that long-acting factors may provide a new paradigm in hemophilia management, with prolonged protection against spontaneous bleeds, decreased infusion frequency, and improved quality of life. An unanswered question is whether individual pharmacokinetics will be necessary to personalize or optimize dosing of long-acting clotting factors or if empiric dosing, as is commonly practiced today, will be sufficient to provide maximal bleed protection. Although cost and patient choice may also shape discussions regarding implementation of these products, the new pharmacokinetic characteristics of long-acting proteins offer the potential to reduce disease burden and improve quality with fewer treatments, constituting a major improvement in hemophilia management.

It is important to recognize that, whereas long-acting factor proteins have significant potential to reduce burden of disease, considerable efforts continue to be spent on the development of alternative modalities for the treatment of hemophilia and other congenital disorders. Although the current strategy of regular factor infusions to prevent bleeds (ie, prophylaxis) reduces morbidity and improves quality of life, the most difficult treatment complication, inhibitor formation, remains in up to 30% of patients. Therefore, better approaches are still needed. The newest approach, in contrast to infusing factor proteins with prolonged factor half-life, is to block clot inhibitors, thereby enhancing thrombin generation and hemostasis. Some of these novel prohemostatic approaches are in early-phase studies and are beginning to be developed for phase 3 trials in individuals with hemophilia and other congenital bleeding disorders. These include inhibitors of RNA interference silencing of antithrombin,60  monoclonal anti-TFPI antibody,61  anti-TFPI aptamer,62  bispecific anti-IXa/X antibody,63  and fucoidans.64  If proven safe and effective in phase 3 trials, these novel agents may provide for long-lasting, inhibitor-free therapy for hemophilia because they avoid exposure of hemophilia patients to standard factor VIII or factor IX to which inhibitor formation is directed, a potentially game-changing innovation.

Conflict-of-interest disclosure: The author is on the board of directors or an advisory committee for Biogen Idec and National Hemophilia Foundation Medical and Scientific Advisory Committee; has received research funding from Baxter, Bayer, Biogen Idec, CSL Behring, and Novo Nordisk; and has consulted for Baxter and Biogen Idec. Off-label drug use: PCCs and rFVIIa for reversal of NOACs.

Margaret V. Ragni, MD, MPH, Professor of Medicine, Clinical and Translational Science, Department of Medicine, Division Hematology/Oncology, University of Pittsburgh, Director, Hemophilia Center of Western Pennsylvania, 3636 Boulevard of the Allies, Pittsburgh, PA 15213-4306; Phone: 412-209-7288; Fax: 412-209-7281; e-mail: ragni@dom.pitt.edu.

1
Tanaka
 
KA
Key
 
NS
Levy
 
JH
Blood coagulation: hemostasis and thrombin regulation
Anesth Analg
2009
, vol. 
108
 
5
(pg. 
1433
-
1446
)
2
Tanaka
 
KA
Taketomi
 
T
Szlam
 
F
Calatzis
 
A
Levy
 
JH
Improved clot formation by combined administration of activated factor VII (NovoSeven) and fibrinogen (Haemocomplettan P)
Anesth Analg
2008
, vol. 
106
 
3
(pg. 
732
-
738
)
3
Skolnick
 
BE
Mathews
 
DR
Khutoryansky
 
NM
Pusateri
 
AE
Carr
 
ME
Exploratory study on the reversal of warfarin with rFVIIa in healthy subjects
Blood
2010
, vol. 
116
 
5
(pg. 
693
-
701
)
4
Yank
 
V
Tuohy
 
CV
Logan
 
AC
, et al. 
Systematic review: benefits and harms of in- hospital use of recombinant factor VIIa for off-label indications
Ann Intern Med
2011
, vol. 
154
 
8
(pg. 
529
-
540
)
5
Logan
 
AC
Yank
 
V
Stafford
 
RS
Off-label use of recombinant factor VIIa in U.S. hospitals: analysis of hospital records
Ann Intern Med
2011
, vol. 
154
 
8
(pg. 
516
-
522
)
6
Fukuda
 
T
Honda
 
Y
Kamisato
 
C
, et al. 
Reversal of anticoagulant effects of edoxaban, an oral direct factor Xa inhibitor, with haemostatic agents
Thromb Haemost
2012
, vol. 
107
 
2
(pg. 
253
-
259
)
7
Mitterlechner
 
T
Innerhofer
 
P
Streif
 
W
, et al. 
Prothrombin complex concentrate and recombinant prothrombin alone or in combination with recombinant factor X and FVIIa indilutional coagulopathy: a porcine model
J Thromb Haemost
2011
, vol. 
9
 
4
(pg. 
729
-
737
)
8
Dickniete
 
G
Dorr
 
B
Kaspereit
 
F
Tanaka
 
KA
Prothrombin complex concentrate versus recombinant factor VIIa for reversal of hemodilutional coagulopathy in a porcine trauma model
J Trauma
2010
, vol. 
68
 
5
(pg. 
1151
-
1157
)
9
Taketomi
 
T
Szlam
 
F
Bader
 
SO
Sheppard
 
CA
Levy
 
JH
Tanaka
 
KA
Effects of recombinant activated factor VII on thrombin-mediated feedback activation of coagulation
Blood Coagul Fibrinolysis
2008
, vol. 
19
 
2
(pg. 
135
-
141
)
10
Holbrook
 
A
Schulman
 
S
Witt
 
DM
, et al. 
Evidence-based management of anticoagulant therapy: antithrombotic therapy and prevention of thrombosis. 9th Ed. American College of Chest Physicians
Chest
2012
, vol. 
141
 
Suppl 1
(pg. 
e152S
-
e184S
)
11
Pabinger
 
I
Brenner
 
B
Kalina
 
U
, et al. 
Beriplex P/N anticoagulation reversal study group. Prothrombin complex concentrate (Beriplex P/N) for emergency anticoagulation reversal: a prospective multinational clinical trial
J Thromb Haemost
2008
, vol. 
6
 
4
(pg. 
622
-
631
)
12
Tanaka
 
KA
Mazzeffi
 
MA
Brube
 
M
Ogawa
 
S
Chen
 
EP
Three-factor prothrombin complex concentrate and hemostasis after high-risk cardiovascular surgery
Transfusion
2013
, vol. 
53
 
4
(pg. 
920
-
921
)
13
Sniecinski
 
RM
Chen
 
EP
Levy
 
JH
Szlam
 
F
Tanaka
 
KA
Coagulopathy after cardiopulmonary bypass in Jehovah's Witness patients: management of two cases using fractionated components and factor VIIa
Anesth Analg
2007
, vol. 
104
 
4
(pg. 
763
-
765
)
14
Schick
 
KS
Fertmann
 
JM
Jauch
 
KW
Hoffman
 
JN
Prothrombin complex concentrate in surgical patients: retrospective evaluation of Vitamin K antagonist reversal and treatment of severe bleeding
Crit Care
2009
, vol. 
13
 pg. 
R191
 
15
Gill
 
R
Herbertson
 
M
Vuylsteke
 
A
, et al. 
Safety and efficacy of recombinant activated factor VII: a randomized placebo-controlled trial in the setting of bleeding after cardiac surgery
Circulation
2009
, vol. 
120
 
1
(pg. 
21
-
27
)
16
Joseph
 
B
Amini
 
A
Friese
 
RS
, et al. 
Factor IX complex for the correction of traumatic coagulopathy
J Trauma Acute Care Surg
2012
, vol. 
72
 
4
(pg. 
828
-
834
)
17
Spinella
 
PC
Perkins
 
JG
McLaughlin
 
DF
, et al. 
The effect of recombinant activated factor VII on mortality in combat-related casualties with severe trauma and massive transfusion
J Trauma
2008
, vol. 
64
 
2
(pg. 
286
-
293
)
18
Joseph
 
B
Hadjizacharia
 
P
Aziz
 
H
, et al. 
Prothrombin complex concentrate: an effective therapy in reversing the coagulopathy of traumatic brain injury
J Trauma Acute Care Surg
2013
, vol. 
74
 
1
(pg. 
248
-
253
)
19
Sarode
 
R
Matevosyan
 
K
Bhagat
 
R
, et al. 
Rapid warfarin reversal: a 3-factor prothrombin complex concentrate and recombinant factor VIIa cocktail for intracerebral hemorrhage
J Neurosurg
2012
, vol. 
116
 
3
(pg. 
491
-
497
)
20
CSL Behring
An open-label, randomized, multicenter phase IIIb study to assess the efficacy, safety, and tolerance of Beriplex P/N compared with plasma for rapid reversal of coagulopathy induced by vitamin K antagonists in subjects requiring an urgent surgical procedure
Accessed July 15, 2013 
21
Weber
 
CF
Gorlinger
 
K
Meininger
 
D
, et al. 
Point-of-care testing: a prospective, randomized clinical trial of efficacy in coagulopathic cardiac surgery patients
Anesthesiology
2012
, vol. 
117
 
3
(pg. 
531
-
547
)
22
Ogawa
 
S
Szlam
 
F
Ohnishi
 
T
Molinaro
 
RJ
Hosokawa
 
K
Tanaka
 
KA
A comparative study of prothrombin complex concentrates and fresh frozen plasma for warfarin reversal under static and flow conditions
Thromb Haemost
2011
, vol. 
106
 
6
(pg. 
1215
-
1223
)
23
Holland
 
L
Warkentin
 
TE
Refaai
 
M
, et al. 
Suboptimal effect of a three-factor prothrombin complex concentrate (Profilnine-SD) in correcting supratherapeutic international normalized ratio due to warfarin overdose
Transfusion
2009
, vol. 
49
 
6
(pg. 
1171
-
1177
)
24
Riess
 
HB
Meier-Hellmann
 
A
Motsch
 
J
Elias
 
M
Kursten
 
FW
Dempfle
 
CE
Prothrombin complex concentrate (Octaplex) in patients requiring immediate reversal of oral anticoagulation
Thromb Res
2007
, vol. 
121
 
1
(pg. 
9
-
16
)
25
Leissinger
 
CA
Blatt
 
PM
Hoots
 
WK
Ewenstein
 
B
Role of prothrombin complex concentrates in reversing warfarin anticoagulation: a review of the literature
Am J Hematol
2008
, vol. 
83
 
2
(pg. 
137
-
143
)
26
Imberti
 
D
Barillari
 
G
Biasioli
 
C
, et al. 
Emergency anticoagulation reversal with 3-factor prothrombin complex concentrates in patients with intracranial hemorrhage
Blood Transfus
2011
, vol. 
9
 
2
(pg. 
148
-
155
)
27
Makris
 
M
van Veen
 
JJ
Three or four factor prothrombin complex concentrates for emergency anticoagulation reversal?
Blood Transfus
2011
, vol. 
9
 
2
(pg. 
117
-
119
)
28
Rosovsky
 
RP
Crowther
 
MA
What is the evidence for the off-label use of recombinant factor VIIa (rFVIIa) in the acute reversal of warfarin?
Hematology Am Soc Hematol Educ Program
2008
, vol. 
2008
 (pg. 
36
-
38
)
29
Sniecinski
 
RM
Chen
 
EP
Makadia
 
SS
Kikura
 
M
Bolliger
 
D
Tanaka
 
KA
Changing from aprotinin to tranexamic acid results in increased use of blood products and recombinant factor VIIa for aortic surgery requiring hypothermic arrest
J Cardiothorac Vasc Anesth
2010
, vol. 
24
 
6
(pg. 
959
-
963
)
30
Marlu
 
R
Hodaj
 
E
Paris
 
A
Albaladejo
 
P
Crackowski
 
JL
Pernod
 
G
Effect of nonspecific reversal agents on anticoagulant activity of dabigatran and rivaroxaban: a randomized crossover ex vivo study in healthy volunteers
Thromb Haemost
2012
, vol. 
108
 
2
(pg. 
217
-
224
)
31
Lambourne
 
MD
Eltringham-Smith
 
LJ
Gataince
 
S
, et al. 
Prothrombin complex concentrates reduce blood loss in murine coagulopathy induced by warfarin, but not in that induced by dabigatran etexilate
J Thromb Haemost
2012
, vol. 
10
 
9
(pg. 
1830
-
1840
)
32
Pragst
 
I
Zeitler
 
SH
Doerr
 
B
, et al. 
Reversal of dabigatran anticoagulation by prothrombin complex concentrate (Beriplex P/N) in a rabbit model
J Thromb Haemost
2012
, vol. 
10
 
9
(pg. 
1841
-
1848
)
33
Zhou
 
W
Schwarting
 
S
Illanes
 
S
, et al. 
Hemostatic therapy in experimental intracerebral hemorrhage associated with the direct thrombin inhibitor dabigatran
Stroke
2011
, vol. 
42
 
12
(pg. 
3594
-
3599
)
34
Perzborn
 
E
Bruber
 
A
Tinel
 
H
, et al. 
Reversal of rivaroxaban anticoagulation by haemostatic agents in rats and primates
Thromb Haemost
2013
, vol. 
110
 
1
(pg. 
162
-
172
)
35
Eerenberg
 
E
Kamphuisen
 
P
Sijpkens
 
M
Meijers
 
JC
Buller
 
HR
Levi
 
M
Reversal of rivaroxaban and dabigatran by prothrombin complex concentrate: a randomized, placebo- controlled, crossover study in healthy subjects
Circulation
2011
, vol. 
124
 
14
(pg. 
1573
-
1579
)
36
Warkentin
 
TE
Margetts
 
P
Connolly
 
SJ
Lamy
 
A
Ricci
 
C
Eikelboom
 
JW
Recombinant VIIa (rFVIIa) and hemodialysis to manage massive dabigatran-associated postcardiac surgery bleeding
Blood
2012
, vol. 
119
 
9
(pg. 
2172
-
2174
)
37
Levy
 
JH
Tanaka
 
KA
Dietrich
 
W
Perioperative hemostatic management of patients treated with vitamin K antagonists
Anesthesiology
2008
, vol. 
109
 
5
(pg. 
918
-
926
)
38
Weitz
 
JI
Quinlan
 
DJ
Eikelboom
 
JW
Periprocedural management and approach to bleeding in patients taking dabigatran
Circulation
2012
, vol. 
126
 
20
(pg. 
2428
-
2432
)
39
Levi
 
M
Eerenberg
 
E
Kamphuise
 
PW
Bleeding risk and reversal strategies for old and new anticoagulants and antiplatelet agents
J Thromb Haemost
2011
, vol. 
9
 
9
(pg. 
1705
-
1712
)
40
Baglin
 
T
Hillarp
 
A
Tripodi
 
A
Elalamy
 
I
Buller
 
H
Ageno
 
W
Measuring Oral Direct Inhibitors (ODIs) of thrombin and factor Xa: A recommendation from the Subcommittee on Control of Anticoagulation of the Scientific and Standardisation Committee of the International Society on Thrombosis and Haemostasis
J Thromb Haemost
2013
, vol. 
11
 
4
(pg. 
756
-
760
)
41
van Ryn
 
J
Sieger
 
P
Kink-Eiband
 
M
Gansser
 
D
Clemens
 
A
Adsorption of dabigatran etexilate in water or dabigatran in pooled human plasma by activated charcoal in vitro [abstract]
Blood (ASH Annual Meeting Abstracts)
2009
, vol. 
114
 
22
pg. 
1065
 
42
Ghanny
 
S
Warkentin
 
TE
Crowther
 
MA
Reversing anticoagulation therapy
Curr Drug Discov Technol
2012
, vol. 
9
 
2
(pg. 
143
-
149
)
43
van Ryn
 
J
Schurer
 
J
Kink-Eiband
 
M
Clemens
 
A
The successful reversal of dabigatran-induced bleeding by coagulation factor concentrates in a rat tail bleeding model do not correlate with ex vivo markers of anticoagulation [abstract]
Blood (ASH Annual Meeting Abstracts)
2011
, vol. 
118
 
21
pg. 
2316
 
44
Kaatz
 
S
Kouides
 
PA
Garcia
 
DA
, et al. 
Guidance on the emergent reversal of oral thrombin and factor Xa inhibitors
Am J Hematol
2012
, vol. 
87
 
S1
(pg. 
S141
-
S145
)
45
Lu
 
G
DeGuzman
 
F
Lakhotia
 
S
, et al. 
Recombinant antidote for reversal of anticoagulation by factor Xa inhibitors [abstract]
Blood (ASH Annual Meeting Abstracts)
2008
, vol. 
112
 
11
pg. 
983
 
46
Tanaka
 
KA
Gruber
 
A
Szlam
 
F
, et al. 
Interaction between thrombin mutant W215A/ E217A and direct thrombin inhibitor
Blood Coagul Fibrinolysis
2008
, vol. 
19
 
5
(pg. 
465
-
467
)
47
Schiele
 
F
van Ryn
 
J
Canada
 
K
, et al. 
A specific antidote for dabigatran: functional and structural characterization
Blood
2013
, vol. 
121
 
18
(pg. 
3554
-
3562
)
48
Kalina
 
U
Bickhard
 
H
Schulte
 
S
Biochemical comparison of seven commercially available prothrombin complex concentrates
Int J Clin Pract
2008
, vol. 
62
 
10
(pg. 
1614
-
1622
)
49
Barillari
 
G
Pasca
 
SM
Bariollari
 
A
De Angelis
 
V
Emergency reversal of anticoagulation: from theory to real use of prothrombin complex concentrates. A retrospective Italian experience
Blood Transfus
2012
, vol. 
10
 
1
(pg. 
87
-
94
)
50
Demeyere
 
R
Gillardin
 
S
Arnout
 
J
Strengers
 
PF
Comparison of fresh frozen plasma and prothrombin complex concentrates for the reversal of oral anticoagulants in patients undergoing cardiopulmonary bypass surgery: A randomized study
Vox Sang
2010
, vol. 
99
 
3
(pg. 
251
-
260
)
51
Wojick
 
C
Schymik
 
ML
Cure
 
EG
Activated prothrombin complex concentrate factor VIII inhibitor bypass activity (FEIBA) for the reversal of warfarin-induced coagulopathy
Int J Emerg Med
2009
, vol. 
2
 
4
(pg. 
217
-
225
)
52
Negrier
 
C
Knobe
 
K
Tiede
 
A
Giangrande
 
P
Møss
 
J
Enhanced pharmacokinetic properties of a glycoPEGylated recombinant factor IX: a first human dose trial in patients with hemophilia B
Blood
2011
, vol. 
118
 
10
(pg. 
2695
-
2701
)
53
Shapiro
 
A
Ragni
 
MV
Valentino
 
LA
, et al. 
Recombinant factor IX-Fc fusion protein (rFIXFc) demonstrates safety and prolonged activity in a phase 1/2a study in hemophilia B patients
Blood
2012
, vol. 
119
 
2
(pg. 
666
-
672
)
54
Santagastino
 
E
Negrier
 
C
Klamroth
 
R
, et al. 
Safety and pharmacokinetics of a novel recombinant fusion protein linking coagulation factor IX with albumin (rIX-FP) in hemophilia B patients
Blood
2012
, vol. 
120
 
12
(pg. 
2405
-
2411
)
55
Powell
 
J
Ozelo
 
M
Pasi
 
J
, et al. 
Results from a phase 3 study of safety, efficacy, and pharmacokinetics of long-lasting recombinant factor IX Fc fusion protein (rFIXFc)
Paper presented at the XXIV Congress of the International Society on Thrombosis and Haemostasis (ISTH)
July 3, 2013
Amsterdam
56
Metzner
 
HJ
Weimer
 
T
Kronthaler
 
U
, et al. 
Genetic fusion to albumin improves the pharmacokinetic properties of factor IX
Thromb Haemost
2009
, vol. 
102
 
4
(pg. 
634
-
644
)
57
Peters
 
RT
Low
 
SC
Kamphaus
 
GD
, et al. 
Prolonged activity of factor IX as a monomeric Fc Fusion protein
Blood
2010
, vol. 
115
 
10
(pg. 
2057
-
2064
)
58
Roopenian
 
DC
Akilesh
 
S
FcRn: The neonatal Fc receptor comes of age
Nat Rev Immunol
2007
, vol. 
7
 
9
(pg. 
715
-
725
)
59
Dumont
 
JA
Liu
 
T
Low
 
SC
, et al. 
Prolonged activity of a recombinant factor VIII Fc fusion protein in hemophilia A mice and dogs
Blood
2012
, vol. 
119
 
13
(pg. 
3024
-
3030
)
60
Akinc
 
A
Sehgal
 
A
Barros
 
S
, et al. 
An RNAi therapeutic targeting antithrombin increases thrombin generation in nonhuman primates [abstract]
Blood (ASH Annual Meeting Abstracts)
2012
, vol. 
120
 
21
pg. 
3370
 
61
Hilden
 
I
Lauiritzen
 
B
Sorensen
 
BB
, et al. 
Hemostatic effect of a monocloncal antibody mAB 2021 blocking the interaction between FXa and TFPI in a rabbit hemophilia model
Blood
2012
, vol. 
119
 
24
(pg. 
5871
-
5878
)
62
Kitazawa
 
T
Igawa
 
T
Sampei
 
Z
, et al. 
A bispecific antibody to factors IXa and X restores factor VIII hemostatic activity in a hemophilia A model
Nat Med
2012
, vol. 
18
 
10
(pg. 
1570
-
1574
)
63
Goyczyca
 
ME
Nair
 
SC
Jilma
 
B
, et al. 
Inhibition of tissue factor pathway inhibitor by the aptamer Bax499 improves clotting of hemophilic blood and plasma
J Thromb Haemost
2012
, vol. 
10
 
8
(pg. 
1581
-
1590
)
64
Zhao
 
X
Dong
 
S
Wang
 
J
, et al. 
A comparative study of antithrombotic and antiplatelet activities of different fucoidans from Laminaria japonica
Thromb Res
2012
, vol. 
129
 
6
(pg. 
771
-
778
)